EP1104310A1 - Expression et caracterisation d'une proteine d'enveloppe vih-1 associee a une reponse d'anticorps de neutralisation largement reactifs - Google Patents

Expression et caracterisation d'une proteine d'enveloppe vih-1 associee a une reponse d'anticorps de neutralisation largement reactifs

Info

Publication number
EP1104310A1
EP1104310A1 EP99940868A EP99940868A EP1104310A1 EP 1104310 A1 EP1104310 A1 EP 1104310A1 EP 99940868 A EP99940868 A EP 99940868A EP 99940868 A EP99940868 A EP 99940868A EP 1104310 A1 EP1104310 A1 EP 1104310A1
Authority
EP
European Patent Office
Prior art keywords
hiv
proteins
dna
amino acid
neutralization
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
EP99940868A
Other languages
German (de)
English (en)
Other versions
EP1104310A4 (fr
EP1104310B1 (fr
Inventor
Gerald Dep. Preventive Med. & Biometrics QUINNAN
Peng Fei Dep. Preventive Med. & Biometrics ZHANG
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Henry M Jackson Foundation for Advancedment of Military Medicine Inc
Original Assignee
Henry M Jackson Foundation for Advancedment of Military Medicine Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Henry M Jackson Foundation for Advancedment of Military Medicine Inc filed Critical Henry M Jackson Foundation for Advancedment of Military Medicine Inc
Publication of EP1104310A1 publication Critical patent/EP1104310A1/fr
Publication of EP1104310A4 publication Critical patent/EP1104310A4/fr
Application granted granted Critical
Publication of EP1104310B1 publication Critical patent/EP1104310B1/fr
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5256Virus expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • C12N2740/16122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to HIV-1 envelope proteins and peptides derived from the donor of the Neutralizing Reference Human Serum (2) which is noted for its capacity to neutralize primary HIV isolates of varied subtypes.
  • NA HIV-1 neutralizing antibodies
  • variable neutralization domains include those in variable (V) regions 1, 2, and 3 of gpl20, while the conserved domains include the primary receptor binding site, and other epitopes in gpl20 and gp41.
  • Amino acid sequence variation is undoubtedly the explanation for the variation that is seen in specificity of neutralization sensitivity among virus strains.
  • the present inventors have cloned and characterized the envelope genes from the donor of human serum which is noted for its capacity to neutralize primary HIV isolates of various subtype (Vujcic, et al. 1995, D'Souza et al, 1991).
  • the invention includes an isolated HIV envelope protein or fragment thereof which, when injected into a mammal, induces the production of broadly cross-reactive neutralizing anti-serum against multiple strains of HIV-1.
  • the invention further includes an isolated HIV envelope protein or fragment thereof comprising a pro line at a position corresponding to amino acid residue 313, a methionine at a position corresponding to amino acid residue 314 and a glutamine at a position corresponding to amino acid residue 325 of SEQ ID NO:l.
  • the invention includes an isolated HIV envelope glycoprotein or fragment thereof comprising an alanine at a position corresponding to amino acid residue 167 of SEQ ID NO:l.
  • the invention also includes an isolated HIV envelope protein comprising the amino acid sequence of SEQ ID NO:l as well as an isolated nucleic acid molecule encoding the envelope protein.
  • compositions for eliciting an immune response such as vaccines, immunogenic compositions and attenuated viral vaccine delivery vectors comprising the envelope proteins, peptides and nucleic acids encoding such proteins and peptides of the invention are also included.
  • Methods for generating antibodies in a mammal comprising administering one or more of these proteins, peptides and nucleic acids, in an amount sufficient to induce the production of the antibodies, is also included in the invention.
  • the invention also comprises a diagnostic reagent comprising one or more of the isolated HIV-1 envelope proteins and methods for detecting broadly cross-reactive neutralizing anti-serum against multiple strains of HIV-1.
  • Figure 1 Phylogenetic analysis of the gpl20 and gp41 nucleotide coding sequences of clone R2. Alignments were performed using the Clustial algorithm of Higgins and Sharp in the program DNA Star (Higgins et al, 1989; Saitou et al, 1987; Myers et al, 1988). The graphs at the bottom of the two figures indicate the percent similarity distances represented by the dendograms.
  • Gene bank accession numbers for the sequences represented are: MW 959, U08453; MW960, U08454; D747, X65638; BR020, U27401; BR029, U27413; RU131, U30312; UG975, U27426; AD8, M60472; HXB, K03455; NDK, M27323; Z2Z6, M22639; UG021, U27399; CM235, L03698; TH022, U09139; TH006, U08810; UG275, L22951; SF1703, M66533; RW020, U08794; RW009, U08793; U455, M62320; and Z321, M15896.
  • FIG. 2 Neutralization of clade B viruses and pseudoviruses by sera from 10 male residents of the Baltimore/Washington, D.C. area collected from 1985-1990 in the Multicenter AIDS Cohort Study.
  • the P9 and PI 0 viruses (P9-V and P10-V) are primary isolates from two of the serum donors (Quinnan et al, 1998).
  • the neutralization assays were performed in PM1 cells, as described in the Examples. Each point represents the results obtained with an individual serum.
  • the open bars represent the standard deviations about the geometric means, indicated by the midlines.
  • the numbers above the results obtained using pseudoviruses indicate the probabilities obtained from testing the null hypothesis by paired t testing comparing the individual pseudoviruses to R2.
  • the neutralization endpoints for 90% neutralization were calculated as described previously (Quinnan et al, 1999; Quinnan et al, 1998; Zhang et al, 1999; Park et al, 1998). Results shown are means of triplicate determinations.
  • the peptide concentrations are 3 x 10 raised to the indicated power.
  • Figure 3 Comparative inhibitory effects of peptides on neutralization of R2 and MN (clone V5) pseudoviruses. All peptides were tested at 15 ⁇ g/ml.
  • the linear peptides (L) corresponded to the apical sequences of the respective V3 loops.
  • the cyclic peptides (C) corresponded to the full lengths of the respective V3 regions of the different strains. Neutralization in the absence of peptide (None), is also shown.
  • NA neutralizing antibody
  • the present inventors have studied envelope protein from a donor with non-progressive HIV-1 infection whose serum contains broadly cross-reactive, primary virus NA. DNA was extracted from lymphocytes, which had been collected approximately six and twelve months prior to the time of collection of the cross reactive serum, env genes were synthesized by nested PCR, cloned, expressed on pseudoviruses, and phenotyped in NA assays. Two clones from each time point had identical V3 region nucleotide sequences, utilized CCR5 but not CXCR4 for cell entry, and had similar reactivities with two reference sera.
  • the present invention relates to HIV-1 envelope proteins from this donor who had non-progressive HIV-1 infection whose serum contains broadly cross-reactive, primary virus neutralizing antibody.
  • the invention also relates to isolated or purified proteins and protein fragments that share certain amino acids at particular positions with the foregoing HIV-1 proteins.
  • Proteins and peptides of the invention include the full length envelope protein having the amino acid sequence of Table 3 (SEQ ID NO:l), gpl20 having the amino acid sequence corresponding to gpl20 in Table 3 (amino acids: 1-520 of SEQ ID NO:l), gp41 having the amino acid sequence corresponding to gp41 in Table 3 (amino acids 521-866 of SEQ ID NO:l), as well as polypeptides and peptides corresponding to the V3 domain and other domains such as VI /V2, C3, V4, C4 and V5. These domains correspond to the following amino acid residues of SEQ ID NO:l:
  • Polypeptides and peptides comprising any single domain may be of variable length but include the amino acid residues of Table 3 (SEQ ID NO:l) which differ from previously sequenced envelope proteins.
  • peptides of the invention which include all or part of the V3 domain may comprise the sequence: PM X ! X 2 X 3 X 4 X 5 X 6 X 7 X g X 9 X 10 Q, wherein X ⁇ -X ⁇ 0 are any natural or non-natural amino acids (P refers to Proline, M refers to methionine and Q refers to Glutamine).
  • Non-natural amino acids include, for example, beta-alanine (beta- Ala), or other omega-amino acids, such as 3-amino propionic, 2,3-diamino propionic (2,3-diaP), 4-amino butyric and so forth, alpha-aminisobutyric acid (Aib), sarcosine (Sat), ornithine (Orn), citrulline (Cit), t-butylalanine (t-BuA), t-butylglycine (t-BuG), N-methylisoleucine (N-Melle), phenylglycine (Phg), and cyclohexylalanine (Cha), norleucine (Nle), cysteic acid (Cya) 2-naphthylalanine (2-Nal); l,2,3,4-tetrahydroisoquinoline-3-carboxylic acid (Tic); beta-2-thienylalanine (Thi
  • peptides of the invention are 60%, 70%, 80% or more preferably, 90% identical to the V3 region of the HIV envelope protein of Table 3 (SEQ ID NO:l).
  • V3 peptides of the invention comprise about 13 amino acids but may be 14, 15, 17, 20, 25, 30, 35, 36, 39, 40, 45, 50 or more amino acids in length.
  • a V3 peptide of 13 amino acids in length consists of the sequence PMGPGRAFYTTGQ (amino acids 313-325 of Table 3 (SEQ ID NO: 1).
  • polypeptides and peptides comprising all or part of the VI /V2 domain comprise an amino acid sequence with an alanine residue at a position corresponding to amino acid 167 Table 3 (SEQ ID NO:l).
  • peptides of the invention spanning the V1/V2 domain may comprise the sequence FNIATSIG (residues 164-171 of SEQ ID NO:l) and may be about 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more amino acids in length.
  • a position corresponding to refers to amino acid positions in HIV envelope proteins or peptides of the invention which are equivalent to a given amino acid residue in the sequence of Table 1 (SEQ ID NO:l) in the context of the surrounding residues.
  • the peptides of the present invention may be prepared by any known techniques.
  • the peptides may be prepared using the solid-phase synthetic technique initially described by Merrifield (1965), which is incorporated herein by reference. Other peptide synthesis techniques may be found, for example, in Bodanszky et al, Peptide Synthesis, 2d ed. (New York, Wiley, 1976).
  • Proteins and peptides of the invention may be prepared by any available means, including recombinant expression of the desired protein or peptide in eukaryotic or prokaryotic host cells (see U.S. Patent 5,696,238).
  • Methods for producing proteins or peptides of the invention for purification may employ conventional molecular biology, microbiology, and recombinant DNA techniques within the ordinary skill level of the art. Such techniques are explained fully in the literature. See, for example, Maniatis et al, Molecular Cloning: A Laboratory Manual, 2d ed. (Cold Spring Harbor, Cold Spring Harbor Laboratory Press, 1989); Glover, DNA Cloning: A Practical Approach, Vols. 1-4 (Oxford, IRL Press, 1985); Gait, Oligonucleotide Synthesis: A Practical Approach
  • the present invention further provides nucleic acid molecules that encode the proteins or peptides of the invention. Such nucleic acid molecules can be in an isolated form, or can be operably linked to expression control elements or vector sequences.
  • the present invention further provides host cells that contain the vectors via transformation, transfection, electroporation or any other art recognized means of introducing a nucleic acid into a cell.
  • a "cell line" is a clone of a primary cell that is capable of stable growth in vitro for many generations.
  • a DNA "coding sequence” is a double-stranded DNA sequence which is transcribed and translated into a polypeptide in vivo when placed under the control of appropriate regulatory sequences.
  • the boundaries of the coding sequence are determined by a start codon at the 5' (amino) terminus and a translation stop codon at the 3' (carboxy) terminus.
  • a polyadenylation signal and transcription termination sequence will usually be located 3' to the coding sequence.
  • a "heterologous" region of the DNA construct is an identifiable segment of DNA within a larger DNA molecule that is not found in association with the larger molecule in nature.
  • the gene when the heterologous region encodes a mammalian gene, the gene will usually be flanked by DNA that does not flank the mammalian genomic DNA in the genome of the source organism.
  • Another example of a heterologous coding sequence is a construct where the coding sequence itself is not found in nature (e.g., a cDNA where the genomic coding sequence contains introns, or synthetic sequences having codons different than the native gene). Allelic variations or naturally-occurring mutational events do not give rise to a heterologous region of DNA as defined herein.
  • naked DNA means nucleic acid molecules that are free from viral particles, particularly retroviral particles. This term also means nucleic acid molecules which are free from facilitator agents including but not limited to the group comprising: lipids, liposomes, extracellular matrix-active enzymes, saponins, lectins, estrogenic compounds and steroidal hormones, hydroxylated lower alkyls, dimethyl sulfoxide (DMSO) and urea.
  • facilitator agents including but not limited to the group comprising: lipids, liposomes, extracellular matrix-active enzymes, saponins, lectins, estrogenic compounds and steroidal hormones, hydroxylated lower alkyls, dimethyl sulfoxide (DMSO) and urea.
  • nucleic acid molecule refers to the polymeric form of deoxyribonucleotides (adenine, guanine, thymine, and/or cytosine) in either its single stranded form, or in double-stranded helix as well as RNA. This term refers only to the primary and secondary structure of the molecule and is not limited to any particular tertiary form. In discussing the structure of particular double-stranded DNA molecules, sequences may be described herein according to the normal convention of giving only the sequence in the 5' to 3' direction along the nontranscribed strand of DNA (e.g. , the strand having a sequence homologous to the mRNA).
  • Transcriptional and translational control sequences are DNA regulatory sequences, such as promoters, enhancers, polyadenylation signals, terminators, and the like, that provide for the expression of a coding sequence in a host cell.
  • a "promoter sequence” is a DNA regulatory region capable of binding RNA polymerase in a cell and initiating transcription of a downstream (3' direction) coding sequence.
  • the promoter sequence is bounded (inclusively) at its 3' terminus by the transcription initiation site and extends upstream (5 1 direction) to include the minimum number of bases or elements necessary to initiate transcription at levels detectable above background.
  • a transcription initiation site within the promoter sequence will be found a transcription initiation site, as well as protein binding domains responsible for the binding of RNA polymerase.
  • Eukaryotic promoters will often, but not always, contain "TATA" boxes and "CAT” boxes.
  • a "replicon” is any genetic element (e.g., plasmid, chromosome, virus) that functions as an autonomous unit of DNA replication in vivo; i.e., capable of replication under its own control.
  • a "signal sequence” can be included before the coding sequence or the native 29 amino acid signal sequence from the envelope protein of Table 3 may be used. This sequence encodes a signal peptide, N-terminal to the polypeptide, that communicates to the host cell to direct the polypeptide to the cell surface or secrete the polypeptide into the media. This signal peptide is clipped off by the host cell before the protein leaves the cell. Signal sequences can be found associated with a variety of proteins native to prokaryotes and eukaryotes. For instance, alpha-factor, a native yeast protein, is secreted from yeast, and its signal sequence can be attached to heterologous proteins to be secreted into the media (See U.S. Pat. No.
  • alpha-factor and its analogs have been found to secrete heterologous proteins from a variety of yeast, such as Saccharomyces and Kluyveromyces, (EP 88312306.9; EP 0324274 publication, and EP 0301669).
  • yeast such as Saccharomyces and Kluyveromyces
  • An example for use in mammalian cells is the tPA signal used for expressing Factor VIIIc light chain.
  • DNA sequences are "substantially homologous" when at least about 85% (preferably at least about 90% and most preferably at least about 95%) of the nucleotides match over the defined length of the DNA sequences. Sequences that are substantially homologous can be identified in a Southern hybridization experiment under, for example, stringent conditions as defined for that particular system. Defining appropriate hybridization conditions is within the skill of the art. See, for example, Maniatis et al, supra. A cell has been "transformed" by exogenous or heterologous DNA when such
  • the transforming DNA may or may not be integrated (covalently linked) into chromosomal DNA making up the genome of the cell.
  • the transforming DNA may be maintained on an episomal element such as a plasmid or viral vector.
  • a stably transformed cell is one in which the transforming DNA has become integrated into a chromosome so that it is inherited by daughter cells through chromosome replication. This stability is demonstrated by the ability of the eukaryotic cell to establish cell lines or clones comprised of a population of daughter cells containing the transforming DNA.
  • a coding sequence is "under the control" of transcriptional and translational control sequences in a cell when RNA polymerase transcribes the coding sequence into mRNA, which is then translated into the protein encoded by the coding sequence.
  • a "vector” is a replicon, such as plasmid, phage or cosmid, to which another DNA segment may be attached so as to bring about the replication of the attached segment.
  • Vectors are used to simplify manipulation of the DNA which encodes the HIV proteins or peptides, either for preparation of large quantities of DNA for further processing (cloning vectors) or for expression of the HIV proteins of peptides (expression vectors).
  • Vectors comprise plasmids, viruses (including phage), and integrated DNA fragments, i.e., fragments that are integrated into the host genome by recombination.
  • Cloning vectors need not contain expression control sequences.
  • control sequences in an expression vector include transcriptional and translational control sequences such as a transcriptional promoter, a sequence encoding suitable ribosome binding sites, and sequences which control termination of transcription and translation.
  • the expression vector should preferably include a selection gene to facilitate the stable expression of HIV gene and/or to identify transformants.
  • the selection gene for maintaining expression can be supplied by a separate vector in cotransformation systems using eukaryotic host cells.
  • Suitable vectors generally will contain replicon (origins of replication, for use in non-integrative vectors) and control sequences which are derived from species compatible with the intended expression host.
  • replicable vector as used herein, it is intended to encompass vectors containing such replicons as well as vectors which are replicated by integration into the host genome.
  • Transformed host cells are cells which have been transformed or transfected with vectors containing HIV peptide or protein encoding DNA.
  • the expressed HIV proteins or peptides may be secreted into the culture supernatant, under the control of suitable processing signals in the expressed peptide, e.g. homologous or heterologous signal sequences.
  • Expression vectors for host cells ordinarily include an origin of replication, a promoter located upstream from the HIV protein or peptide coding sequence, together with a ribosome binding site, a polyadenylation site, and a transcriptional termination sequence. Those of ordinary skill will appreciate that certain of these sequences are not required for expression in certain hosts.
  • An expression vector for use with microbes need only contain an origin of replication recognized by the host, a promoter which will function in the host, and a selection gene.
  • promoters are derived from polyoma, bovine papilloma virus, CMV (cytomegalovirus, either murine or human), Rouse sarcoma virus, adenovirus, and simian virus 40 (SV40).
  • Other control sequences e.g., terminator, poly A, enhancer, or amplification sequences
  • terminator e.g., poly A, enhancer, or amplification sequences
  • An expression vector is constructed so that the HIV protein or peptide coding sequence is located in the vector with the appropriate regulatory sequences, the positioning and orientation of the coding sequence with respect to the control sequences being such that the coding sequence is transcribed and translated under the "control" of the control sequences (i.e., RNA polymerase which binds to the DNA molecule at the control sequences transcribes the coding sequence).
  • the control sequences may be ligated to the coding sequence prior to insertion into a vector, such as the cloning vectors described above.
  • the coding sequence can be cloned directly into an expression vector which already contains the control sequences and an appropriate restriction site. If the selected host cell is a mammalian cell, the control sequences can be heterologous or homologous to the HIV coding sequence, and the coding sequence can either be genomic DNA containing introns or cDNA.
  • Higher eukaryotic cell cultures may be used to express the proteins of the present invention, whether from vertebrate or invertebrate cells, including insects, and the procedures of propagation thereof are known. See, for example, Kruse & Patterson,
  • Suitable host cells for expressing HIV proteins or peptides in higher eukaryotes include: monkey kidney CVI line transformed by SV40 (COS-7, ATCC CRL1651); baby hamster kidney cells (BHK, ATCC CRLIO); Chinese hamster ovary-cells-DHFR (Uriaub
  • African green monkey kidney cells (VERO76, ATCC CRL1587); human cervical carcinoma cells (HeLa, ATCC CCL2); canine kidney cells (MDCK, ATCC
  • HIV gene products may have higher molecular weights than expected due to glycosylation. It is therefore intended that partially or completely glycosylated forms of
  • HIV preproteins or peptides having molecular weights somewhat different from 160, 120 or 41 kD are within the scope of this invention.
  • vaccinia virus which is well-known in the art. See, for example, Macket et al. (1984); Glover, supra; and WO
  • Yeast expression vectors are known in the art. See, for example, U.S. Patents
  • vector pHSI transforms Chinese hamster ovary cells
  • Mammalian tissue may be cotransformed with DNA encoding a selectable marker such as dihydrofolate reductase (DHFR) or thymidine kinase and DNA encoding the HIV protein or peptide.
  • DHFR dihydrofolate reductase
  • thymidine kinase DNA encoding the HIV protein or peptide.
  • DHFR dihydrofolate reductase
  • thymidine kinase DNA encoding the HIV protein or peptide.
  • An appropriate host cell in this case is the Chinese hamster ovary (CHO) cell line deficient in DHFR activity, prepared and propagated as described by Uriaub & Chasin, (1980).
  • HIV proteins or peptides are produced by growing host cells transformed by an exogenous or heterologous DNA construct, such as an expression vector described above under conditions whereby the HIV protein is expressed. The HIV protein or peptide is then isolated from the host cells and purified. If the expression system secretes the protein or peptide into the growth media, the protein can be purified directly from cell-free media. The selection of the appropriate growth conditions and initial crude recovery methods are within the skill of the art.
  • a coding sequence for an HIV protein or peptide of the invention can be cloned into any suitable vector and thereby maintained in a composition of cells which is substantially free of cells that do not contain an HIV coding sequence.
  • Numerous cloning vectors are known to those of skill in the art. Examples of recombinant DNA vectors for cloning and host cells which they can transform include the various bacteriophage lambda vectors (E. coli), pBR322 (E. coli), pACYC177 (E. coli), pKT230 (gram-negative bacteria), pGV 1106 (gram-negative bacteria), pLAFRI
  • HIV envelope fusion proteins and methods for making such proteins have been previously described (U.S. Patent 5,885,580). It is now a relatively straight forward technology to prepare cells expressing a foreign gene. Such cells act as hosts and may include, for the fusion proteins of the present invention, yeasts, fungi, insect cells, plants cells or animals cells. Expression vectors for many of these host cells have been isolated and characterized, and are used as starting materials in the construction, through conventional recombinant DNA techniques, of vectors having a foreign DNA insert of interest. Any DNA is foreign if it does not naturally derive from the host cells used to express the DNA insert.
  • the foreign DNA insert may be expressed on extrachromosomal plasmids or after integration in whole or in part in the host cell chromosome(s), or may actually exist in the host cell as a combination of more than one molecular form.
  • the choice of host cell and expression vector for the expression of a desired foreign DNA largely depends on availability of the host cell and how fastidious it is, whether the host cell will support the replication of the expression vector, and other factors readily appreciated by those of ordinary skill in the art.
  • the foreign DNA insert of interest comprises any DNA sequence coding for fusion proteins including any synthetic sequence with this coding capacity or any such cloned sequence or combination thereof.
  • fusion proteins coded and expressed by an entirely recombinant DNA sequence is encompassed by this invention but not to the exclusion of fusion proteins peptides obtained by other techniques.
  • Vectors useful for constructing eukaryotic expression systems for the production of fusion proteins comprise the fusion protein's DNA sequence, operatively linked thereto with appropriate transcriptional activation DNA sequences, such as a promoter and/or operator.
  • appropriate transcriptional activation DNA sequences such as a promoter and/or operator.
  • Other typical features may include appropriate ribosome binding sites, termination codons, enhancers, terminators, or replicon elements. These additional features can be inserted into the vector at the appropriate site or sites by conventional splicing techniques such as restriction endonuclease digestion and ligation.
  • Yeast expression systems which are the preferred variety of recombinant eukaryotic expression system, generally employ Saccharomyces cerevisiae as the species of choice for expressing recombinant proteins.
  • Saccharomyces cerevisiae Other species of the genus Saccharomyces are suitable for recombinant yeast expression system, and include but are not limited to carlsbergensis, uvarum, rouxii, montanus, kluyveri, elongisporus, norbensis, oviformis, and diastaticus.
  • Saccharomyces cerevisiae and similar yeasts possess well known promoters useful in the construction of expression systems active in yeast, including but not limited to GAP, GAL 10, ADH2, PHO5, and alpha mating factor.
  • Yeast vectors useful for constructing recombinant yeast expression systems for expressing fusion proteins include, but are not limited to, shuttle vectors, cosmid plasmids, chimeric plasmids, and those having sequences derived from two micron circle plasmids. Insertion of the appropriate DNA sequence coding for fusion proteins into these vectors will, in principle, result in a useful recombinant yeast expression system for fusion proteins where the modified vector is inserted into the appropriate host cell, by transformation or other means.
  • Recombinant mammalian expression system are another means of producing the fusion proteins for the vaccines/immunogens of this invention.
  • a host mammalian cell can be any cell that has been efficiently cloned in cell culture.
  • mammalian expression options can be extended to include organ culture and transgenic animals.
  • Host mammalian cells useful for the purpose of constructing a recombinant mammalian expression system include, but are not limited to, Vero cells, NIH3T3, GH3, COS, murine C127 or mouse L cells.
  • Mammalian expression vectors can be based on virus vectors, plasmid vectors which may have SV40, BPV or other viral replicons, or vectors without a replicon for animal cells. Detailed discussions on mammalian expression vectors can be found in the treatises of Glover, DNA Cloning: A Practical Approach, Vols. 1-4 (Oxford, IRL Press, 1985).
  • Fusion proteins may possess additional and desirable structural modifications not shared with the same organically synthesized peptide, such as adenylation, carboxylation, glycosylation, hydroxylation, methylation, phosphorylation or myristylation. These added features may be chosen or preferred as the case may be, by the appropriate choice of recombinant expression system. On the other hand, fusion proteins may have its sequence extended by the principles and practice of organic synthesis.
  • the proteins or peptides of the present invention may be used as "subunit" vaccines or immunogens.
  • Such vaccines or immunogens offer significant advantages over traditional vaccines in terms of safety and cost of production; however, subunit vaccines are often less immunogenic than whole-virus vaccines, and it is possible that adjuvants with significant immunostimulatory capabilities may be required in order to reach their full potential.
  • adjuvants approved for human use in the United States include aluminum salts (alum). These adjuvants have been useful for some vaccines including hepatitis B, diphtheria, polio, rabies, and influenza.
  • Other useful adjuvants include Complete Freund's Adjuvant (CFA), Incomplete Freund's Adjuvant (IF A), Muramyl dipeptide (MDP) (see Ellouz et al, 1974), synthetic analogues of MDP (reviewed in Chedid et al, 1978), N-acetylmuramyl-L-alanyl-D-isoglutamyl-L-alanine- 2-[ 1 ,2-dipalmitoyl-s-glycero-3-(hydroxyphosphoryloxy)]ethylamide (MTP-PE) and compositions containing a metabolizable oil and an emulsifying agent, wherein the oil and emulsifying agent are present in the form of an oil-in- water emulsion having oil
  • the formulation of a vaccine or immunogenic compositions of the invention will employ an effective amount of the protein or peptide antigen. That is, there will be included an amount of antigen which, in combination with the adjuvant, will cause the subject to produce a specific and sufficient immunological response so as to impart protection to the subject from subsequent exposure to an HIV virus.
  • the formulation When used as an immunogenic composition, the formulation will contain an amount of antigen which, in combination with the adjuvant, will cause the subject to produce specific antibodies which may be used for diagnostic or therapeutic purposes.
  • the vaccine compositions of the invention may be useful for the prevention or therapy of HIV-1 infection. While all animals that can be afflicted with HIV-1 can be treated in this manner, the invention, of course, is particularly directed to the preventive and therapeutic use of the vaccines of the invention in man. Often, more than one administration may be required to bring about the desired prophylactic or therapeutic effect; the exact protocol (dosage and frequency) can be established by standard clinical procedures.
  • the vaccine compositions are administered in any conventional manner which will introduce the vaccine into the animal, usually by injection.
  • the vaccine composition can be administered in a form similar to those used for the oral administration of other proteinaceous materials.
  • the precise amounts and formulations for use in either prevention or therapy can vary depending on the circumstances of the inherent purity and activity of the antigen, any additional ingredients or carriers, the method of administration and the like.
  • the vaccine dosages administered will typically be, with respect to the gpl20 antigen, a minimum of about 0.1 mg/dose, more typically a minimum of about 1 mg/dose, and often a minimum of about 10 mg/dose.
  • the maximum dosages are typically not as critical. Usually, however, the dosage will be no more than 500 mg/dose, often no more than 250 mg/dose.
  • These dosages can be suspended in any appropriate pharmaceutical vehicle or carrier in sufficient volume to carry the dosage.
  • the final volume, including carriers, adjuvants, and the like typically will be at least 0.1 ml, more typically at least about 0.2 ml.
  • the upper limit is governed by the practicality of the amount to be administered, generally no more than about 0.5 ml to about 1.0 ml.
  • Peptides of the invention corresponding to domains of the envelope protein such as V3 may be constructed or formulated into compounds or compositions comprising multimers of the same domain or multimers of different domains.
  • peptides corresponding to the V3 domain may be circularized by oxidation of the cysteine residues to form multimers containing 1, 2, 3, 4 or more individual peptide epitopes.
  • the circularized form may be obtained by oxidizing the cysteine residues to form disulfide bonds by standard oxidation procedures such as air oxidization.
  • Synthesized peptides of the invention may also be circularized in order to mimic the geometry of those portions as they occur in the envelope protein. Circularization may be facilitated by disulfide bridges between existing cysteine residues. Cysteine residues may also be included in positions on the peptide which flank the portions of the peptide which are derived from the envelope protein. Alternatively, cysteine residues within the portion of a peptide derived from the envelope protein may be deleted and/or conservatively substituted to eliminate the formation of disulfide bridges involving such residues. Other means of circularizing peptides are also well known.
  • vaccine or immunogenic compositions may be prepared as vaccine vectors which express the HIV protein or peptide of the invention in the host animal.
  • Any available vaccine vector may be used, including live Venezuelan Equine Encephalitis virus (see U.S. Patent 5,643,576), poliovirus (see U.S. Patent 5,639,649), pox virus (see U.S. Patent 5,770,211) and vaccina virus (see U.S. Patents 4,603,112 and 5,762,938).
  • naked nucleic acid encoding a protein or peptide of the invention may be administered directly to effect expression of the antigen (see U.S. Patent 5,739,118).
  • HIV protein or peptide compositions of the present invention may be used as diagnostic reagents in immunoassays to detect anti-HIV antibodies, particularly anti-gpl20 antibodies.
  • Many HIV immunoassay formats are available. Thus, the following discussion is only illustrative, not inclusive. See generally, however, U.S. Patents 4,743,678; 4,661,445; and 4,753,873 and EP 0161150 and EP 0216191.
  • Immunoassay protocols may be based, for example, upon composition, direct reaction, or sandwich-type assays. Protocols may also, for example, be heterogeneous and use solid supports, or may be homogeneous and involve immune reactions in solution. Most assays involved the use of labeled antibody or polypeptide.
  • the labels may be, for example, fluorescent, chemiluminescent, radioactive, or dye molecules.
  • Assays which amplify the signals from the probe are also known, examples of such assays are those which utilize biotin and avidin, and enzyme-labeled and mediated immunoassays, such as ELISA assays.
  • an immunoassay for anti-HIV antibody will involve selecting and preparing the test sample, such as a biological sample, and then incubating it with an HIV protein or peptide composition of the present invention under conditions that allow antigen-antibody complexes to form. Such conditions are well known in the art.
  • the protein or peptide is bound to a solid support to facilitate separation of the sample from the polypeptide after incubation.
  • solid supports examples include nitrocellulose, in membrane or microtiter well form, polyvinylchloride, in sheets or microtiter wells, polystyrene latex, in beads or microtiter plates, polyvinlyidine fluoride, diazotized paper, nylon membranes, activated beads, and Protein A beads. Most preferably, Dynatech, Immulon® microtiter plates or 0.25 inch polystyrene beads are used in the heterogeneous format.
  • the solid support is typically washed after separating it from the test sample. In homogeneous format, on the other hand, the test sample is incubated with the
  • HIV protein or peptide in solution, under conditions that will precipitate any antigen-antibody complexes that are formed, as is known in the art.
  • the precipitated complexes are then separated from the test sample, for example, by centrifugation.
  • the complexes formed comprising anti-HIV antibody are then detected by any number of techniques. Depending on the format, the complexes can be detected with labeled anti-xenogenic Ig or, if a competitive format is used, by measuring the amount of bound, labeled competing antibody.
  • Diagnostic probes useful in such assays of the invention include antibodies to the HIV-1 envelope protein.
  • the antibodies to may be either monoclonal or polyclonal, produced using standard techniques well known in the art (See Harlow & Lane,
  • Antibodies A Laboratory Manual, (Cold Spring Harbor, Cold Spring Harbor Laboratory Press, 1988). They can be used to detect HIV-1 envelope protein by specifically binding to the protein and subsequent detection of the antibody-protein complex by ELISA, Western blot or the like.
  • the HIV-1 envelope protein used to elicit these antibodies can be any of the variants discussed above.
  • Antibodies are also produced from peptide sequences of HIV-1 envelope proteins using standard techniques in the art (Harlow & Lane, supra). Fragments of the monoclonals or the polyclonal antisera which contain the immunologically significant portion can also be prepared.
  • Peripheral blood mononuclear cells that had been cryopreserved from donations obtained approximately six months and one year prior to the time of Reference 2 collections were used as sources of DNA for env gene cloning. The cells had not been stored to maintain viability. DNA was extracted using phenol/chloroform from approximately 1-3 x 10 6 cells from each donation (Quinnan et al, 1998). The DNA was used as template in a nested polymerase chain reaction, similar to that described previously, except rTth was used as the DNA polymerase, following the manufacturer's instructions (Barnes, 1992; Cariello et al, 1991). The DNA was cloned into the expression vector pSV7d, as previously described (Quinnan et al, 1998; Stuve et al, 1987).
  • HIV-1 env clones in the expression vector pSV3 were obtained from the AIDS Research and Reference Reagent Program, 93MW965.26 (clade C), 92RW ⁇ 20.5 (clade A), 93TH966.8 (clade E), 92UG975.10 (clade G) (Gao et al, 1994).
  • the production of env clones from the molecular virus clones NL43, AD8, and SF162 has been previously described (Quinnan et al, 1998; Adachi et al, 1986; Theodore et al, 1996; Englund et al, 1995).
  • the H9 cell line was obtained from Robert Gallo (Mann et al, 1989).
  • the Molt 3 cell line was obtained from the American Type Culture Collection, Rockville, MD (ATCC). (Daniel et al, 1988)
  • the HOS cell lines expressing CD4 and various coreceptors for HIV-1 were obtained from the NTH AIDS Research and Reference
  • the 293T cell line was obtained from the ATCC, with permission from the Rockefeller Institute (Liou et al, 1994).
  • the H9, Molt3 and PMl cell cultures were maintained in RPMI-1640 medium supplemented with 10% fetal bovine serum and antibiotics (Gibco).
  • the HOS and 293T cells were maintained in Dulbecco's Minimal Essential Medium (Gibco), with similar supplements, except that the HOS cell medium was supplemented with puromycin for maintenance of plasmid stability.
  • Cryopreserved human peripheral blood lymphocytes were stimulated with PHA and used for virus infections (Quinnan et al, 1998; Mascola et al, 1994).
  • the virus NL43 was used in neutralization assays which employed Molt3 cells as target cells and used giant cell formation for endpoint determination, as previously described (Vujcic et al, 1995). The amounts of virus used were sufficient to result in the formation of 30-50 giant cells per well (Vujcic et al, 1995; Lennette, 1964).
  • the viruses, NL(SF 162) and AD8, P9 and P 10 were tested for neutralization in PHA stimulated human lymphoblasts in the presence of IL-2 (Quinnan et al, 1998; Mascola et al, 1994). In the latter assays ten percent of the cell suspension was removed each week, fifty percent of the medium was changed each week, and medium was sampled twice weekly from each well for reverse transcriptase assay.
  • the reverse transcriptase assays were performed on the test samples from the first sampling date at which the non-neutralized control wells had reverse transcriptase activity about 10-20 x background, generally on day 14 or 17 of the assay.
  • the neutralization endpoint was considered to be the highest dilution of serum at which reverse transcriptase activity was reduced at least fifty percent.
  • the Reference Neutralizing Sera 1 and 2 and the Negative Reference Serum were used as positive and negative controls (NTH AIDS Research and Reference Reagent Program)
  • Pseudoviruses were constructed and assayed using methods similar to those described previously (Quinnan et al, 1998; Deng et al, 1996; Park et al, 1998).
  • pSV7d- env plasmid DNA and pNL43.1uc+.E-R- were cotransfected into 70 to 80% confluent 293T cell cultures using the calcium phosphate/Hepes buffer technique, following manufacturer's instructions (Promega, Madison, WI), in 24 well plastic tissue culture trays or 25 cm 2 flasks (Quinnan et al, 1998; Deng et al, 1996; Park et al, 1998).
  • Pseudovirus infectivity was assayed in PMl or HOS-CD4 cells expressing various co-receptors. Transfection supernatants were serially diluted and inoculated into cells in 96 well plates, 50 ⁇ l per well. Assays were routinely performed in triplicate. The cultures were incubated for four days, centrifuged at 400 x g for ten minutes if PMl cells were used, and medium removed by aspiration.
  • the cells were washed twice with phosphate buffered saline, lysed with 25 ⁇ l cell culture lysing reagent according to the manufacturer's instructions (Promega, Madison, WI); the cells were then tritated into the medium, and 10 ⁇ l of the suspensions were transferred to wells of 96 well luminometer plates. Substrate was added in 100 ⁇ l volumes automatically, and the luminescence read using a MicroLumatPlus luminometer (EG&G Berthold, Hercules, CA).
  • Mock PV controls were used in each assay consisting of media harvested from 293T cell cultures transfected with pSV7d (without an env insert) and pNL43.Luc.E-R-, and processed in the same way as cultures for PV preparation. Infectivity endpoints were determined by a modified Reed Munch method; an individual well was considered positive if the luminescence was at least 10-fold greater than the mock control, and the endpoint was considered to be the highest dilution at which the calculated frequency of positivity was ⁇ 50% (Quinnan et al, 1998; Park et al, 1998; Lennette, 1964). Luminescence resulting from infection with minimally diluted samples was generally about 10,000-fold greater than background. Neutralization tests were performed using PMl or HOS-CD4 cells. Aliquots of
  • the neutralization endpoints were calculated by a modified Reed-Munch method in which the endpoint was considered to be the highest serum dilution calculated to have a frequency of ⁇ 50% for reducing luminescence by ⁇ 90% compared to the non-neutralized control. PV titrations were conducted in duplicate in parallel with each neutralization assay.
  • Nucleotide sequence analysis was performed using the di-deoxy cycle sequencing technique and AmpliTaq FS DNA polymerase, according to manufacturer's directions (Perkin Elmer Applied Biosystems, Foster City, CA). After the sequencing reaction the DNA was purified using Centriflex Gel Filtration Cartridges (Advanced Genetic Technologies, Gaithersberg, MD). Sequencing gels were run and analyzed using an Applied Biosystems Prism, Model 377 DNA Sequencer. Sequencing was performed on both strands. Sequence alignment was performed using the Editseq SEQMAN, and
  • Nucleotide sequences including the V3 regions were analyzed for each clone, with more than 300 bases assigned for each, and no differences between the clones were found (results not shown). Based on the absence of demonstration of differences in these assays, a single clone from the March sample was selected for use in subsequent assays, and is designated R2, hereafter.
  • the complete nucleotide sequence of the env gene clone R2 was determined, and found to have an open reading frame of 2598 bases (Genbank Accession Number: AF128126)
  • the amino acid sequence deduced from this sequence is shown in Table 3 (SEQ ID NO: 1).
  • the consensus glycosylation sequences at residues 215 and 270 are highly and moderately variable, respectively.
  • Genotypic analyses conducted included evaluation of the gpl20 and gp41 nucleotide coding sequences in comparison to those of a number of strains of clades A through G, as shown in Figure 1 (Saitou et al, 1987; Myers & Miller, 1988). Both coding regions were more closely related to clade B than non-clade B sequences. Comparative analyses of regions of the predicted gpl20 and gp41 amino acid sequences were also performed (results not shown).
  • each constant and variable region of gpl20 The regions analyzed included: each constant and variable region of gpl20; the proximal gp41 ectodomain including the leucine zipper region; the part of gp41 extending from the end of the leucine zipper to the second cysteine; the remaining gp41 ectodomain, and the transmembrane region; and the cytoplasmic region.
  • R2 consistently related more closely with the clade B sequences than the others.
  • Example 3 Comparative Sensitivity ofR2 and Other Clade B Viruses and Pseudoviruses to Neutralization by Sera from Individuals with Clade B Infections
  • Example 4 Comparative neutralization of pseudoviruses expressing R2 and other envelopes of diverse subtypes by sera from diverse subtype infections.
  • the frequency of neutralization by sera from individuals infected with different clades was not significantly skewed for any of the other four pseudoviruses.
  • Clade A, C, D and G pseudoviruses were neutralized by eight, seventeen, six and three of the seventeen sera tested, respectively.
  • the clade C pseudovirus was substantially more sensitive to neutralization, in general than the others tested.
  • the clade E pseudovirus was neutralized by five of five clade D sera and seven of eight clade E sera but only one of the sera from people infected by other clades.
  • Example 5 Synthetic peptides generated from V3 amino acid sequences from R2 strain.
  • R2 strain V3 peptides were synthesized using an automated ABI synthesizer and
  • the R2 V3 35-mer was insoluble in water, while all other peptides tested were soluble in water to at least 10 mg/ml.
  • solutions of the R2 and R2(313-4PM/HI, 325Q/D) V3 35-mers in dimethylsulfoxide (DMSO), 10 mg/ml were diluted 1 : 10 in water at room temperature or 37°C and the pH was adjusted to 8.5 with ammonium hydroxide. These solutions were aerated by bubbling air through the solutions for periods > 1 hour. Following aeration, the pH was adjusted to 7.4 using hydrochloric acid. A portion of the R2 35-mer peptide precipitated during these procedures.
  • DMSO dimethylsulfoxide
  • Example 6 Peptide blocking of neutralizing antibody activity against clone R2 pseudovirus.
  • Example 7 Cyclic R2 V3 peptide inhibition of neutralization of R2 pseudoviruses by sera from MACS patients. Inhibition of heterologous serum neutralization of R2 pseudovirus by cyclic R2 V3 peptide was evaluated to determine if cross reactivity of these sera with R2 included effects of anti-V3 antibodies.
  • the comparative neutralization titers of sera from ten patients from the MACS against clone R2 pseudovirus in the presence and absence of cyclic R2 V3 peptide are shown in Figure 4A (Quinnan et al, 1998). These sera have been described previously, and have been shown to neutralize primary HIV-1 enveloped pseudoviruses cross reactively, but to a lesser extent than Reference 2 (Zhang et al, 1999).
  • Example 8 Cyclic R2 V3 peptide inhibition of Reference 2 neutralization of pseudoviruses expressing envelopes from the MACS patients.
  • Example 9 Induction of cross-reactive neutralizing antibodies in mice following immunization with recombinant delivery vectors encoding HIV-1 envelope proteins.
  • the DNA clone encoding the R2 envelope was introduced into an expression vector which can be used to express the envelope protein complex in vivo for immunization.
  • the recombinant delivery vector expressing the R2 envelope clone was been administered to mice, both in its full length, encoding both gpl20 and gp41, or in a truncated form.
  • the truncated form is secreted by cells which express gp 140. Both the full-length and truncated form of these constructs induced neutralizing antibodies in mice.
  • mice which received the g ⁇ l40 construct, which includes the V3 region have developed neutralizing antibodies which neutralize at least three different strains of HIV-1, including the R2 strain, a macrophage tropic laboratory strain known as SF162, and a primary strain which is not laboratory adapted.
  • the amount of cross-reactivity observed exceeds that induced by most or all other HIV immunogens that have been tested as single agents.
  • 'Neutralization titers are the dilutions at which 90% inhibition of luminescence was observed.
  • b Sera were the Reference Neutralizing Human Serum 1 and 2, or were provided by Dr. J. Mascola, HIVNET, or the UNAIDS Program, as described in the text.
  • c NT not tested.
  • Adachi A Gendelman HE, Keonig S, Folks T, Wiley R, Rabson A and Martin MA, Production of acquired immunodeficiency syndrome-associated retrovirus in human and nonhuman cells transfected with an infectious molecular clone. J Virol. (1986) 59: 284-291.
  • Englund G Theodore TS, Freed EO, Engleman A and Martin MA, Integration is required for productive infection of monocyte-derived macrophages by human immunodeficiency virus type 1. J Virol. (1995) 69: 3216-3219.
  • T cells expressing activated LF A- 1 are more susceptible to infection with human immunodeficiency virus type 1 particles bearing host-encoded ICAM. J Virol. (1998) 72: 2105-2112. Gao F, Yue L, Craig S, Thornton CL, Robertson DL, McCutchan FE, Bradac JA,
  • Higgins DG and Sharp PM Fast and sensitive multiple sequence alignments on a microcomputer.
  • Hioe CE, Xu S, Chigurupati P, Burda S, Williams C, Gorny MK and Zolla-Pazner S Neutralization of HIV-1 primary isolates by polyclonal and monoclonal human antibodies. Int Immunol. (1997) 9: 1281-1290.
  • Lennette EH "General principles underlying laboratory diagnosis of viral and rickettsial infections" in: Lennette EH and Schmidt MJ, Diagnostic Procedures of Viral and Rickettsial Disease (New York, American Public Health Association, 1964) pp. 45-
  • Lusso P Cocchi F, Balotta C, Markham PD, Louie A, Farci P, Pal R, Gallo RC and Reitz MS, Growth ofmacrophage-tropic and primary human immunodeficiency virus tupe 1 (HIV-1) isolates in a unique CD4+ T-cell clone (PMl): failure to downregulate CD4 and to interfere with cell-line HIV-1. J Virol. (1995) 69: 3712-3720.
  • Thali M Furman C, Ho DD, Robinson J, Tilley S, Pinter A and Sodroski J, Discontinuous, conserved neutralization epitopes overlapping the CD4-binding region of human immunodeficiency virus type 1 gpl20 envelope glycoprotein. J Virol. (1992) 66: 5635-5641.
  • Uriaub G and Chasin LA Isolation of Chinese hamster cell mutants deficient in dihydrofolate reductase activity. Proc Natl Acad Sci USA (1980) 77: 4216-4220.
  • VanCott TC VanCott TC
  • Mascola JR Kaminski RW
  • Kalyanaraman V Hallberg PL
  • Burnett PR Ulrich JT
  • Irish DJ and Birx DL Antibodies with specificity to native gpl20 and neutralization activity against primary human immunodeficiency virus type 1 isolates elicited by immunization with oligomeric gpl60.
  • J Virol. (1997) 71 : 4319-4330.
  • Vujcic LK and Quinnan GV Preparation and characterization of human HIV type 1 neutralizing reference sera. (1995) AIDS Res Hum Retroviruses. 11 : 783-787.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Gastroenterology & Hepatology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oncology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Communicable Diseases (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Animal Behavior & Ethology (AREA)
  • AIDS & HIV (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Medicines Containing Plant Substances (AREA)

Abstract

La présente invention concerne des protéines d'enveloppe VIH-1 provenant d'un donneur atteint d'une infection à VIH-1 non progressive, le sérum de ce donneur contenant des anticorps de neutralisation d'un virus primaire largement réactifs. Cette invention concerne également des protéines isolées ou purifiées ainsi que des fragments protéiques partageant des acides aminés avec lesdites protéines VIH-1 sur certaines positions.
EP99940868A 1998-08-04 1999-08-04 Expression et caracterisation d'une proteine d'enveloppe vih-1 associee a une reponse d'anticorps de neutralisation largement reactifs Expired - Lifetime EP1104310B1 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US9526798P 1998-08-04 1998-08-04
US95267P 1998-08-04
PCT/US1999/017596 WO2000007631A1 (fr) 1998-08-04 1999-08-04 Expression et caracterisation d'une proteine d'enveloppe vih-1 associee a une reponse d'anticorps de neutralisation largement reactifs

Publications (3)

Publication Number Publication Date
EP1104310A1 true EP1104310A1 (fr) 2001-06-06
EP1104310A4 EP1104310A4 (fr) 2003-01-08
EP1104310B1 EP1104310B1 (fr) 2009-07-01

Family

ID=22251048

Family Applications (1)

Application Number Title Priority Date Filing Date
EP99940868A Expired - Lifetime EP1104310B1 (fr) 1998-08-04 1999-08-04 Expression et caracterisation d'une proteine d'enveloppe vih-1 associee a une reponse d'anticorps de neutralisation largement reactifs

Country Status (7)

Country Link
EP (1) EP1104310B1 (fr)
JP (1) JP4637356B2 (fr)
AT (1) ATE435034T1 (fr)
AU (1) AU762376C (fr)
CA (1) CA2338886C (fr)
DE (1) DE69941051D1 (fr)
WO (1) WO2000007631A1 (fr)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW511388B (en) 2000-03-28 2002-11-21 Koninkl Philips Electronics Nv Passive radiator having mass elements
CA2483870A1 (fr) * 2002-05-10 2004-06-03 The Henry M. Jackson Foundation Site d'interaction avec la gp41 sur la gp120 du vih-1
JP4898434B2 (ja) * 2003-06-20 2012-03-14 シーメンス・ヘルスケア・ダイアグノスティックス・プロダクツ・ゲーエムベーハー B型肝炎ウィルスの新規の表面タンパク質(HBsAg)変異体
CA2529997A1 (fr) 2003-06-20 2004-12-29 Dade Behring Marburg Gmbh Nouvelle variante de la proteine de surface (hbsag-) du virus de l'hepatite b
GB0417494D0 (en) 2004-08-05 2004-09-08 Glaxosmithkline Biolog Sa Vaccine
JP2009536653A (ja) * 2006-05-09 2009-10-15 ザ ヘンリー エム. ジャクソン ファウンデーション フォー ザ アドヴァンスメント オブ ミリタリー メディシン インコーポレイテッド Hiv−1免疫原性組成物
JP2011502521A (ja) * 2007-11-12 2011-01-27 ザ ヘンリー エム. ジャクソン ファウンデーション フォー ザ アドヴァンスメント オブ ミリタリー メディシン インコーポレイテッド Hiv−1外被糖タンパク質オリゴマー及び使用方法
EA201290956A1 (ru) 2010-03-26 2013-04-30 Глаксосмитклайн Байолоджикалс С.А. Вакцина против вич
CN103228294A (zh) 2010-09-27 2013-07-31 葛兰素史密斯克莱生物公司 疫苗
WO2014039746A1 (fr) 2012-09-07 2014-03-13 Emory University Compositions de stimulation immunitaire du vih comprenant des pili exprimés de façon recombinante sur des bactéries, et procédés associés

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992020373A1 (fr) * 1991-05-14 1992-11-26 Repligen Corporation Anticorps d'heteroconjugues pour le traitement des infections a l'hiv
WO1993005812A1 (fr) * 1991-09-18 1993-04-01 The Government Of The United States Of America As Represented By The Department Of Health And Human Services Procede de production de lymphocytes t cytotoxiques specifiques a une gamme importante d'isolats de hiv-1 au moyen de peptides hybrides synthetiques

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8923123D0 (en) * 1989-10-13 1989-11-29 Connaught Lab A vaccine for human immunodeficiency virus
CA2096159C (fr) * 1992-05-14 2004-07-20 Hermann Katinger Peptides induisant des anticorps qui neutralisent les isolats de vih-1 divergents sur le plan genetique
GB9318901D0 (en) * 1993-09-13 1993-10-27 Centre Nat Rech Scient Multiple branch peptide construction

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992020373A1 (fr) * 1991-05-14 1992-11-26 Repligen Corporation Anticorps d'heteroconjugues pour le traitement des infections a l'hiv
WO1993005812A1 (fr) * 1991-09-18 1993-04-01 The Government Of The United States Of America As Represented By The Department Of Health And Human Services Procede de production de lymphocytes t cytotoxiques specifiques a une gamme importante d'isolats de hiv-1 au moyen de peptides hybrides synthetiques

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
BARBAS III C ET AL: "In vitro evolution of a neutralizing human antibody to human immunodeficiency virus type 1 to enhance affinity and broaden strain cross-reactivity" PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, NATIONAL ACADEMY OF SCIENCE. WASHINGTON, US, vol. 91, no. 9, 26 April 1994 (1994-04-26), pages 3809-3813, XP002111656 ISSN: 0027-8424 *
DATABASE EMBL [Online] 1 August 1998 (1998-08-01) retrieved from EBI Database accession no. O71260 XP002220280 *
DATABASE EMBL [Online] 1 May 1997 (1997-05-01) retrieved from EBI Database accession no. P88523 XP002220281 *
DATABASE EMBL [Online] 1 November 1996 (1996-11-01) retrieved from EBI Database accession no. Q73297 XP002220282 *
DOUGLAS NIGEL W ET AL: "An efficient method for the rescue and analysis of functional HIV-1 env genes: Evidence for recombination in the vicinity of the tat/rev splice site." AIDS (PHILADELPHIA), vol. 10, no. 1, 1996, pages 39-46, XP009000382 ISSN: 0269-9370 *
PURTSCHER MARTIN ET AL: "A broadly neutralizing human monoclonal antibody against gp41 of human immunodeficiency virus type 1." AIDS RESEARCH AND HUMAN RETROVIRUSES, vol. 10, no. 12, 1994, pages 1651-1658, XP001059097 ISSN: 0889-2229 *
QUINNAN GERALD V JR ET AL: "Expression and characterization of HIV type 1 envelope protein associated with a broadly reactive neutralizing antibody response." AIDS RESEARCH AND HUMAN RETROVIRUSES, vol. 15, no. 6, 10 April 1999 (1999-04-10), pages 561-570, XP002220279 ISSN: 0889-2229 *
See also references of WO0007631A1 *
TRKOLA ALEXANDRA ET AL: "Cross-clade neutralization of primary isolates of human immunodeficiency virus type 1 by human monoclonal antibodies and tetrameric CD4-IgG." JOURNAL OF VIROLOGY, vol. 69, no. 11, 1995, pages 6609-6617, XP002220278 ISSN: 0022-538X *

Also Published As

Publication number Publication date
CA2338886A1 (fr) 2000-02-17
ATE435034T1 (de) 2009-07-15
AU5464299A (en) 2000-02-28
CA2338886C (fr) 2011-10-04
AU762376B2 (en) 2003-06-26
JP2002522039A (ja) 2002-07-23
AU762376C (en) 2004-10-14
DE69941051D1 (de) 2009-08-13
EP1104310A4 (fr) 2003-01-08
EP1104310B1 (fr) 2009-07-01
WO2000007631A9 (fr) 2000-11-16
WO2000007631A1 (fr) 2000-02-17
JP4637356B2 (ja) 2011-02-23

Similar Documents

Publication Publication Date Title
Bures et al. Immunization with recombinant canarypox vectors expressing membrane-anchored glycoprotein 120 followed by glycoprotein 160 boosting fails to generate antibodies that neutralize R5 primary isolates of human immunodeficiency virus type 1
AU729231B2 (en) Synthetic HIV genes
AP1282A (en) HIV envelope polypeptides and vaccine.
US20100015211A1 (en) Combination Approaches For Generating Immune Responses
US6585979B1 (en) HIV envelope polypeptides and immunogenic composition
AU762376C (en) Expression and characterization of HIV-1 envelope protein associated with a broadly reactive neutralizing antibody response
US20070166784A1 (en) Combination approaches for generating immune responses
US7608688B2 (en) HIV-1 protein associated with broadly reactive neutralizing antibody response
JP5290576B2 (ja) 修飾されたhiv−1エンベロープタンパク質
AU2003299489B2 (en) Interacting site for gp41 on gp120 of HIV-1
US20030087225A1 (en) Synthetic HIV genes
ZA200603029B (en) Combination approaches for generating immune responses

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20010222

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

A4 Supplementary search report drawn up and despatched

Effective date: 20021126

AK Designated contracting states

Kind code of ref document: A4

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

17Q First examination report despatched

Effective date: 20030929

17Q First examination report despatched

Effective date: 20030929

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: THE HENRY M. JACKSON FOUNDATION FOR THE ADVANCEMEN

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REF Corresponds to:

Ref document number: 69941051

Country of ref document: DE

Date of ref document: 20090813

Kind code of ref document: P

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090701

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090701

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20091012

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090701

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20091102

Ref country code: MC

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20090831

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090701

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20100406

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20090804

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20091002

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090701

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20090804

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090701

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20140827

Year of fee payment: 16

Ref country code: NL

Payment date: 20140826

Year of fee payment: 16

Ref country code: DE

Payment date: 20140827

Year of fee payment: 16

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20140827

Year of fee payment: 16

Ref country code: FR

Payment date: 20140818

Year of fee payment: 16

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: BE

Payment date: 20140829

Year of fee payment: 16

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 69941051

Country of ref document: DE

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20150804

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150831

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150831

REG Reference to a national code

Ref country code: NL

Ref legal event code: MM

Effective date: 20150901

REG Reference to a national code

Ref country code: FR

Ref legal event code: ST

Effective date: 20160429

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150901

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160301

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150804

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150831

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150831